• Non ci sono risultati.

7-Acylamino-3H-1,2-benzoxathiepine 2,2-dioxides as new isoform-selective carbonic anhydrase IX and XII inhibitors

N/A
N/A
Protected

Academic year: 2021

Condividi "7-Acylamino-3H-1,2-benzoxathiepine 2,2-dioxides as new isoform-selective carbonic anhydrase IX and XII inhibitors"

Copied!
8
0
0

Testo completo

(1)

Full Terms & Conditions of access and use can be found at

https://www.tandfonline.com/action/journalInformation?journalCode=ienz20

Journal of Enzyme Inhibition and Medicinal Chemistry

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ienz20

7-Acylamino-3H-1,2-benzoxathiepine 2,2-dioxides

as new isoform-selective carbonic anhydrase IX

and XII inhibitors

Aleksandrs Pustenko , Alessio Nocentini , Anastasija Balašova , Mikhail

Krasavin , Raivis Žalubovskis & Claudiu T. Supuran

To cite this article:

Aleksandrs Pustenko , Alessio Nocentini , Anastasija Balašova , Mikhail

Krasavin , Raivis Žalubovskis & Claudiu T. Supuran (2020) 7-Acylamino-3H-1,2-benzoxathiepine

2,2-dioxides as new isoform-selective carbonic anhydrase IX and XII inhibitors, Journal of Enzyme

Inhibition and Medicinal Chemistry, 35:1, 650-656, DOI: 10.1080/14756366.2020.1722658

To link to this article: https://doi.org/10.1080/14756366.2020.1722658

© 2020 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.

Published online: 21 Feb 2020.

Submit your article to this journal Article views: 579

View related articles View Crossmark data

(2)

RESEARCH PAPER

7-Acylamino-3H-1,2-benzoxathiepine 2,2-dioxides as new isoform-selective

carbonic anhydrase IX and XII inhibitors

Aleksandrs Pustenko

a,b

, Alessio Nocentini

c

, Anastasija Bala

sova

a

, Mikhail Krasavin

d

, Raivis 

Zalubovskis

a,b

and

Claudiu T. Supuran

c

a

Latvian Institute of Organic Synthesis, Riga, Latvia;bInstitute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia;cDipartimento Neurofarba, Sezione di Scienze Farmaceutiche e Nutraceutiche, Universita degli Studi di Firenze, Florence, Italy;dDepartment of Chemistry, Saint Petersburg State University, Saint Petersburg, Russian Federation

ABSTRACT

A series of 3H-1,2-benzoxathiepine 2,2-dioxides incorporating 7-acylamino moieties were obtained by an original procedure starting from 5-nitrosalicylaldehyde, which was treated with propenylsulfonyl chloride followed by Wittig reaction of the bis-olefin intermediate. The new derivatives, belonging to the homosul-focoumarin chemotype, were assayed as inhibitors of the zinc metalloenzyme carbonic anhydrase (CA, EC 4.2.1.1). Four pharmacologically relevant human (h) isoforms were investigated, the cytosolic hCA I and II and the transmembrane, tumour-associated hCA IX and XII. No relevant inhibition of hCA I and II was observed, whereas some of the new derivatives were effective, low nanomolar hCA IX/XII inhibitors, mak-ing them of interest for investigations in situations in which the activity of these isoforms is overex-pressed, such as hypoxic tumours, arthritis or cerebral ischaemia.

ARTICLE HISTORY Received 26 December 2019 Revised 21 January 2020 Accepted 23 January 2020 KEYWORDS Carbonic anhydrase; transmembrane isoforms; sulfocoumarin; homosulfo-coumarin; isoform-selective inhibitor

1. Introduction

Sulfocoumarins (1,2-benzoxathiine 2,2-dioxides) and homosulfo-coumarins (3H-1,2-benzoxathiepine 2,2-dioxides)1–5 are among the most investigated new classes of carbonic anhydrase (CA, EC 4.2.1.1) inhibitors, which have been designed considering the structurally similar coumarins6–8as lead molecules. Indeed, Cas are widely spread enzymes in organisms of all types, from simple to complex ones9–15, and are involved in crucial physiological proc-esses, among which carbon fixation in diatoms and other marine organisms in which several genetic families of such metalloen-zymes were reported9. In protozoans, Cas are involved in biosyn-thetic reactions9 whereas in bacteria, where at least three genetic families were described (a-, b-, and c-Cas) these enzymes play cru-cial roles related both to metabolism but also virulence and sur-vival in various niches10. In vertebrates, including humans, a high number of different CA isoforms belonging to the a-CA class were described11,12, which by hydrating CO2to a weak base (bicarbon-ate) and a strong acid (hydronium ions), are involved in a multi-tude of processes, starting with pH regulation and ending with metabolism13,14. As thus, Cas are drug targets for decades, with their inhibitors having pharmacological applications in a multitude of fields11–16. The primary sulphonamides were discovered as CA inhibitors (CAIs) in the ‘40 s, and most of the drugs that were launched in the next decades as diuretics, antiepileptics, or anti-glaucoma agents belonged to this class of compounds or to their isosteres such as the sulfamates and sulfamides11. An important drawback of such first generation CA inhibitors (CAIs) was their lack of isoform selectivity, considering the fact that in humans at

least 12 catalytically active and three acatalytic isoforms are pre-sent11,12. However, the new generation CAIs to which coumarins

and sulfocoumarins belong, show significant isoform-selective inhibition profiles, as demonstrated in a considerable number of studies1–8. This is principally due to the fact that these com-pounds possess a distinct inhibition mechanism compared to the sulphonamides, which coordinate to the zinc ion from the CA active site as anions11,12. In fact, coumarins and sulfocoumarins act as prodrug inhibitors, undergoing an active site mediated hydrolysis, which leads to the formation of 2-hydroxy-cinnamic acids in the case of the coumarins, and ethane-sulphonates in the case of the sulfocoumarins, which subsequently bind in different active site regions, different of those where the classical sulphona-mide CAIs bind1–8. As shown by X-ray crystallography, the hydro-lysed coumarins occlude the entrance of the CA active site cavity6, whereas the sulfocoumarins bind deeper within the active

site, but still do not coordinate to the metal ion. Instead, the formed sulphonates anchor to the zinc-coordinated water mol-ecule, as shown again by means of X-ray crystallographic techni-ques2. As these regions of the CA active site are the most variable ones, a straightforward explanation of the isoform selectivity of these new generation CAIs was furnished by using a combination of crystallographic and kinetic studies, which also allowed the development of compounds showing a higher degree of selectiv-ity15,16. This allowed for the development of inhibitors useful for new pharmacological applications such as antitumor/antimeta-static compounds13, CAIs useful for the management of arthritis17, neuropathic pain18, and cerebral ischaemia19.

CONTACT Raivis Zalubovskis raivis@osi.lv Latvian Institute of Organic Synthesis, 21 Aizkraukles Str, Riga, LV-1006, Latvia; Claudiu T. Supuran

claudiu.supuran@unifi.it Dipartimento Neurofarba, Sezione di Scienze Farmaceutiche e Nutraceutiche, Universita degli Studi di Firenze, Sesto Fiorentino,

Florence, Italy

ß 2020 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

2020, VOL. 35, NO. 1, 650–656

(3)

Considering our interest in designing non-sulphonamide CAIs with various potential applications, we report here a new series of homosulfocoumarins and their inhibitory profiles against the major human (h) CA isoforms, hCA I, II, IX, and XII, involved in many pathologies, including cancer.

2. Experimental part

2.1. Chemistry

Reagents, starting materials/intermediates 1–7 and solvents were obtained from commercial sources (Sigma-Aldrich, St. Louis, MO) and used as received. Anhydrous CH2Cl2 and toluene were obtained by passing commercially available solvents through acti-vated alumina columns. Thin-layer chromatography was per-formed on silica gel, spots were visualised with UV light (254 and 365 nm). Melting points were determined on an OptiMelt auto-mated melting point system. IR spectra were recorded on Shimadzu FTIR IR Prestige-21 spectrometer. NMR spectra were recorded on Bruker Avance Neo (400 MHz) spectrometer with chemical shifts values (d) in ppm relative to TMS using the residual DMSO-d6 signal (1H 2.50; 13C 39.52) or CDCl3 signal (1H 7.26; 13C 77.16) as an internal standard. High-resolution mass spectra (HRMS) were recorded on a mass spectrometer with a Q-TOF micro mass analyser using the ESI technique.

General procedure for synthesis of acyl compound 8–17

To a solution of amino derivative 7 (1.0 eq.) in dry CH2Cl2 (20 ml per mmol of compound 7) at 0C appropriate acyl chloride (1.1 eq.) and Net3 (1.1 eq.) were added. The resulting mixture was stirred at room temperature under an argon atmosphere for 2 h. Water was added (20 ml per mmol of compound 7). Layers were separated, water layer was washed with EtOAc (2 40 ml). Combined organic layers were washed with brine, dried over anh. Na2SO4, filtered, evaporated. The crude solids were recrystallised form EtOAc/petrol ether mixture to afford product.

N-(2,2-Dioxido-3H-1,2-benzoxathiepin-7-yl)acetamide (8).

O SO2

H N O

Compound 8a was prepared according to the general procedure from amino deriva-tive 7 (150 mg; 0.71 mmol), acetyl chloride (56mL; 0.78 mmol) and Et3N (110mL; 0.78 mmol) as white solid (127 mg; 70%). Mp 164–165C.

IR (film, cm1) max¼ 3276 (N–H), 1670 (C ¼ O), 1370 (S ¼ O), 1361 (S¼ O), 1166 (S ¼ O), 1162 (S ¼ O);

1H NMR (400 MHz, DMSO-d 6) d¼ 2.06 (s, 3H), 4.37–4.41 (m, 2H), 5.96–5.6.04 (m, 1H), 6.89 (d, 1H, J ¼ 11.3 Hz), 7.28 (d, 1H, J ¼ 8.9 Hz), 7.58 (dd, 1H, J ¼ 8.9, 2.5 Hz), 7.69 (d, 1H, J ¼ 2.5 Hz), 10.16 (s, 1H) ppm. 13 C NMR (100 MHz, DMSO-d6) d¼ 24.0, 51.0, 120.6, 120.8, 122.7, 128.4, 131.5, 138.0, 142.2, 168.6 ppm.

HRMS (ESI) [Mþ H]þ: m/z calcd for (C11H12NO4S) 254.0487. Found 254.0498. N-(2,2-Dioxido-3H-1,2-benzoxathiepin-7-yl)benzamide (9). O O SO2 H N

Compound 9 was prepared according to the general procedure from amino derivative 7 (150 mg; 0.71 mmol), ben-zoyl chloride (90mL; 0.78 mmol) and

Et3N (110mL; 0.78 mmol) as white solid (162 mg; 72%). Mp 174–175C.

IR (film, cm1) max¼ 3289 (N–H), 1652 (C ¼ O), 1370 (S ¼ O), 1363 (S¼ O), 1163 (S ¼ O); 1 H NMR (400 MHz, DMSO-d6) d¼ 4.43 (dd, 2H, J ¼ 6.0, 0.9 Hz), 5.99–6.06 (m, 1H), 6.93 (d, 1H, J ¼ 11.2 Hz), 7.35 (d, 1H, J ¼ 8.8 Hz), 7.52–7.58 (m, 2H), 7.59–7.64 (m, 1H), 7.82 (dd, 1H, J ¼ 8.8, 2.5 Hz), 7.91 (d, 1H,J ¼ 2.5 Hz), 7.94–7.99 (m, 2H), 10.46 (s, 1H) ppm. 13 C NMR (100 MHz, DMSO-d6) d¼ 51.1, 120.9, 122.0, 122.1, 122.6, 127.7, 128.3, 128.5, 131.4, 131.8, 134.6, 137.9, 142.7, 165.7 ppm

HRMS (ESI) [Mþ H]þ: m/z calcd for (C16H14NO4S) 316.0644. Found 316.0654. N-(2,2-Dioxido-3H-1,2-benzoxathiepin-7-yl)-4-methy benzamide (10). O SO2 H N O

Compound 10 was prepared according to the general procedure from amino derivative 7 (150 mg; 0.71 mmol), 4-methylbenzoyl chlor-ide (103mL; 0.78 mmol) and Et3N (110mL; 0.78 mmol) as white crys-tals (170 mg; 73%). Mp 197–198C.

IR (film, cm1) max ¼ 3324 (N–H), 1646 (C ¼ O), 1378 (S ¼ O), 1363 (S¼ O), 1177 (S ¼ O), 1169 (S ¼ O);

1 H NMR (400 MHz, DMSO-d6) d¼ 2.39 (s, 3H), 4.41–4.45 (m, 2H), 5.99–6.06 (m, 1H), 6.92 (d, 1H, J ¼ 11.2 Hz), 7.32–7.37 (m, 3H), 7.82 (dd, 1H,J ¼ 8.9, 2.6 Hz), 7.86–7.92 (m, 3H), 10.37 (s, 1H) ppm 13 C NMR (100 MHz, DMSO-d6) d¼ 21.0, 51.1, 120.8, 121.9, 122.1, 122.6, 127.7, 128.3, 129.0, 131.4, 131.6, 138.0, 141.9, 142.6, 165.5 ppm

HRMS (ESI) [Mþ H]þ: m/z calcd for (C17H16NO4S) 330.0800. Found 330.0815. N-(2,2-Dioxido-3H-1,2-benzoxathiepin-7-yl)4-bromobenzamide (11). O O Br SO2 H N

Compound 11 was prepared according to the general proced-ure from amino derivative 7 (150 mg; 0.71 mmol), 4-bromoben-zoyl chloride (171 mg; 0.78 mmol) and Et3N (110mL; 0.78 mmol) as white solid (166 mg; 59%). Mp 185–186C.

IR (film, cm1) max¼ 3260 (N–H), 1653 (C ¼ O), 1375 (S ¼ O), 1363 (S¼ O), 1167 (S ¼ O); 1H NMR (400 MHz, DMSO-d 6) d¼ 4.42–4.46 (m, 2H), 5.99–6.06 (m, 1H), 6.92 (d, 1H,J ¼ 11.3 Hz), 7.35 (d, 1H, J ¼ 8.8 Hz), 7.74–7.83 (m, 3H), 7.88–7.94 (m, 3H), 10.52 (s, 1H) ppm 13 C NMR (100 MHz, DMSO-d6) d¼ 51.2, 120.9, 122.0, 122.2, 122.7, 125.6, 128.3, 129.8, 131.4, 131.5, 133.6, 137.7, 142.8, 164.7 ppm

HRMS (ESI) [Mþ H]þ: m/z calcd for (C16H13BrNO4S) 393.9749. Found 393.9736. N-(2,2-Dioxido-3H-1,2-benzoxathiepin-7-yl)-2-iodobenzamide (12). O SO2 H N O I

Compound 12 was prepared according to the general procedure from amino derivative 7 (150 mg; 0.71 mmol), 2-iodobenzoyl chloride (208 mg; 0.78 mmol) and Et3N (110mL; 0.78 mmol) as white solid (276 mg; 88%). Mp 188–189C.

IR (film, cm1) max¼ 3240 (N–H), 1641 (C ¼ O), 1374 (S ¼ O), 1362 (S¼ O), 1156 (S ¼ O);

(4)

1 H NMR (400 MHz, DMSO-d6) d¼ 4.41–4.45 (m, 2H), 6.00–6.08 (m, 1H), 6.94 (d, 1H, J ¼ 11.2 Hz), 7.22–7.28 (m, 1H), 7.36 (d, 1H, J ¼ 8.8 Hz), 7.47–7.55 (m, 2H), 7.72 (dd, 1H, J ¼ 8.8, 2.5 Hz), 7.87 (d, 1H,J ¼ 2.5 Hz), 7.9–7.97 (m, 1H), 10.67 (s, 1H) ppm 13 C NMR (100 MHz, DMSO-d6) d¼ 51.0, 93.6, 121.0, 121.2, 121.3, 122.9, 128.1, 128.2, 128.5, 131.2, 131.5, 137.7, 139.1, 142.7, 142.8, 167.7 ppm

HRMS (ESI) [Mþ H]þ: m/z calcd for (C16H13INO4S) 441.9610 Found 441.9609. N-(2,2-Dioxido-3H-1,2-benzoxathiepin-7-yl)-2-bromobenzamide (13). O SO2 H N O Br

Compound 13 was prepared accord-ing to the general procedure from amino derivative 7 (150 mg; 0.71 mmol), 2-bromobenzoyl chloride (102mL; 0.78 mmol) and Et3N (110mL; 0.78 mmol) as white solid (230 mg; 82%). Mp 177–178C.

IR (film, cm1) max¼ 3288 (N–H), 1653 (C ¼ O), 1371 (S ¼ O), 1176 (S¼ O), 1156 (S ¼ O); 1 H NMR (400 MHz, DMSO-d6) d¼ 4.43 (dd, 2H, J ¼ 6.0, 0.9 Hz), 6.00–6.07 (m, 1H), 6.94 (d, 1H, J ¼ 11.2 Hz), 7.36 (d, 1H, J ¼ 8.9 Hz), 7.41–7.47 (m, 1H), 7.51 (dt, 1H, J ¼ 7.4, 1.1 Hz), 7.55–7.59 (m, 1H), 7.69–7.76 (m, 2H), 7.87 (d, 1H, J ¼ 2.6 Hz), 10.73 (s, 1H) ppm 13C NMR (100 MHz, DMSO-d 6) d¼ 51.0, 118.9, 121.1, 121.2, 122.9, 127.8, 128.6, 128.9, 131.4, 131.5, 132.8, 137.6, 138.8, 142.8, 166.0 ppm

HRMS (ESI) [Mþ H]þ: m/z calcd for (C16H13BrNO4S) 393.9749 Found 393.9766. N-(2,2-Dioxido-3H-1,2-benzoxathiepin-7-yl)-2-fluorobenzamide (14). O O SO 2 H N F

Compound 14 was prepared accord-ing to the general procedure from amino derivative 7 (150 mg; 0.71 mmol), 2-fluorobenzoyl chloride (93mL; 0.78 mmol) and Et3N (110mL; 0.78 mmol) as white solid (188 mg; 79%). Mp 173–174C.

IR (film, cm1) max¼ 1671 (C ¼ O), 1371 (S ¼ O), 1365 (S ¼ O), 1164 (S¼ O) 1156 (S ¼ O); 1 H NMR (400 MHz, DMSO-d6) d¼ 4.41–4.45 (m, 2H), 6.00–6.07 (m, 1H), 6.93 (d, 1H,J ¼ 11.2 Hz), 7.32–7.40 (m, 3H), 7.56–7.63 (m, 1H), 7.65–7.71 (m, 1H), 7.74 (dd, 1H, J ¼ 8.8, 2.5 Hz), 7.86 (d, 1H, J ¼ 2.5 Hz), 10.65 (s, 1H) ppm 1. 13C NMR (100 MHz, DMSO-d6) d¼ 51.1, 116.2 (d, J ¼ 21.7 Hz), 121.0, 121.4, 121.5, 122.8, 124.6 (d, J ¼ 5.5 Hz), 124.7 (d, J ¼ 6.3 Hz), 128.5, 129.9 (d, J ¼ 2.6 Hz), 131.4, 132.8 (d, J ¼ 8.5 Hz), 137.5, 142.8, 159.9 (d, J ¼ 249 Hz), 163.0 ppm 2. HRMS (ESI) [Mþ H]þ: m/z calcd for (C16H13FNO4S) 334.0549

Found 334.0554. N-(2,2-Dioxido-3H-1,2-benzoxathiepin-7-yl)-2-(trifluoromethyl)ben-zamide (15). O SO2 H N O CF3

Compound 15 was prepared accord-ing to the general procedure from amino derivative 7 (150 mg; 0.71 mmol), 2-(trifluoromethyl)ben-zoyl chloride (115mL; 0.78 mmol) and Et3N (110mL; 0.78 mmol) as white solid (236 mg; 87%). Mp 192–193C.

IR (film, cm1) max¼ 3195 (N–H), 1666 (C ¼ O), 1377 (S ¼ O), 1316 (S¼ O), 1164 (S ¼ O); 1 H NMR (400 MHz, DMSO-d6) d¼ 4.42–4.45 (m, 2H), 6.00–6.08 (m, 1H), 6.94 (d, 1H,J ¼ 11.2 Hz), 7.36 (d, 1H, J ¼ 8.8 Hz), 7.67–7.76 (m, 3H), 7.78–7.89 (m, 3H), 10.81 (s, 1H) ppm 13 C NMR (100 MHz, DMSO-d6) d¼ 51.0, 121.1, 121.3, 122.9, 123.8 (q, J ¼ 274 Hz), 125.8 (q, J ¼ 31.2 Hz), 126.4 (q, J ¼ 4.6 Hz), 128.5, 128.6, 130.3, 131.4, 132.7, 135.8 (q, J ¼ 2.3 Hz), 137.6, 142.8, 165.8 ppm

HRMS (ESI) [Mþ H]þ: m/z calcd for (C17H13NO4 F3S) 384.0517 Found 384.0519. N-(2,2-Dioxido-3H-1,2-benzoxathiepin-7-yl)thiophene-2-carboxa-mide (16). O SO2 H N O S

Compound 16 was prepared accord-ing to the general procedure from amino derivative 7 (150 mg; 0.71 mmol), 2-thiophenecarbonyl chloride (84mL; 0.78 mmol) and Et3N (110mL; 0.78 mmol) as white solid (185 mg; 81%). Mp 162–163C.

IR (film, cm1) max¼ 3357 (N–H), 1648 (C ¼ O), 1372 (S ¼ O), 1356 (S¼ O), 1178 (S ¼ O), 1165 (S ¼ O);

1 H NMR (400 MHz, DMSO-d6) d¼ 4.44 (dd, 2H, J ¼ 6.0, 1.1 Hz), 5.99–6.06 (m, 1H), 6.92 (d, 1H, J ¼ 11.2 Hz), 7.23–7.26 (m, 1H), 7.35 (d, 1H, J ¼ 8.8 Hz), 7.78 (dd, 1H, J ¼ 8.8, 2.6 Hz), 7.84 (d, 1H, J ¼ 2.6 Hz), 7.88 (dd, 1H, J ¼ 5.0, 1.1 Hz), 8.04 (dd, 1H, J ¼ 3.8, 1.1 Hz), 10.43 (s, 1H) ppm 13 C NMR (100 MHz, DMSO-d6) d¼ 51.2, 120.9, 121.9, 122.1, 122.7, 128.2, 128.4, 129.5, 131.3, 132.3, 137.5, 139.5, 142.7, 160.0 ppm

HRMS (ESI) [Mþ H]þ: m/z calcd for (C14H12NO4S2) 322.0208 Found 322.0221. N-(2,2-Dioxido-3H-1,2-benzoxathiepin-7-yl)furan-2-carboxamide (17). O SO2 H N O O

Compound 17 was prepared according to the general procedure from amino derivative 7 (150 mg; 0.71 mmol), 2-furoyl chloride (84mL; 0.78 mmol) and Et3N (110mL; 0.78 mmol) as white solid (185 mg; 81%). Mp 162–163C.

IR (film, cm1) max¼ 3299 (N–H), 1663 (C ¼ O), 1367 (S ¼ O), 1363 (S¼ O), 1165 (S ¼ O), 1158 (S ¼ O);

1 H NMR (400 MHz, DMSO-d6) d¼ 4.41–4.45 (m, 2H), 5.98–6.06 (m, 1H), 6.70–6.74 (m, 1H), 6.90 (d, 1H, J ¼ 11.2 Hz), 7.32–7.38 (m, 2H), 7.80 (dd, 1H,J ¼ 8.8, 2.6 Hz), 7.87 (d, 1H, J ¼ 2.6 Hz), 7.95–7.97 (m, 1H), 10.41 (s, 1H) ppm 13 C NMR (100 MHz, DMSO-d6) d¼ 51.2, 112.3, 115.2, 120.9, 122.0, 122.2, 122.6, 128.3, 131.4, 137.3, 142.7, 146.0, 147.2, 156.3 ppm

HRMS (ESI) [Mþ H]þ: m/z calcd for (C14H12NO5S) 306.0436 Found 306.0463.

2.2. CA inhibitory assay

An applied photophysics stopped-flow instrument has been used for assaying the CA catalysed CO2hydration activity

20

. Phenol red (at a concentration of 0.2 mM) was used as indicator, working at the absorbance maximum of 557 nm, with 20 mM Hepes (pH 7.5) as buffer and 20 mM Na2SO4 (for maintaining constant the ionic strength), following the initial rates of the CA-catalysed CO2 hydra-tion reachydra-tion for a period of 10 100 s. The CO2 concentrations ranged from 1.7 to 17 mM for the determination of the kinetic parameters and inhibition constants. For each inhibitor, at least six traces of the initial 5 10% of the reaction have been used for

(5)

determining the initial velocity. The uncatalysed rates were deter-mined in the same manner and subtracted from the total observed rates. Stock solutions of inhibitor (0.1 mM) were pre-pared in distilled deionised water, and dilutions up to 0.01 nM were done thereafter with the assay buffer. Inhibitor and enzyme solutions were preincubated together for 6 h at room temperature prior to assay in order to allow for the formation of the E I com-plex. The inhibition constants were obtained by nonlinear least-squares methods using PRISM 3 and the Cheng Prusoff equa-tion, as reported earlier21–23, and represent the mean from at least three different determinations. All CA isoforms were recombinant ones obtained in-house as reported earlier21,24.

3. Results and discussion

3.1. Chemistry

Starting from the benzaldehyde derivative 1, the synthesis of the key intermediate 7 was reported earlier by our groups1. Briefly,

the synthesis of 7-amino-3H-1,2-benzoxathiepine 2,2-dioxide (7) was started with a Wittig reaction in which 5-nitro-salicylic alde-hyde 1 was converted to the corresponding mono-olefin 2 in 65% yield (Scheme 1). Treatment of compound 2 with allyl sulphonyl chloride (3) provided the bisolefin 4 in 65% yield. In the next step, the olefin metathesis reaction with Ru-catalyst 5 was employed, leading to the conversion of compound 4 to 7-nitro-3H-1,2-benzoxathiepine 2,2-dioxide 6 in 96% yield. The nitro derivative 6 was thereafter reduced with iron in acidic medium to

the corresponding amine 7 in nearly quantitative yield (98%). The key intermediate 7 was subsequently reacted with a series of acyl chlorides to afford the desired compounds 8–17 in good to Table 1. Inhibition data of human CA isoforms CA I, II, IX and XII with 3H-1,2-benzoxathiepines 2,2-dioxide 8–17 using acetazolamide (AAZ) as a standard drug. O S OO 8-17 H N R O Cmpd R KI(nM) a,b

hCA I hCA II hCA IX hCA XII 8 CH3 >100 mM >100 mM 61.8 162.5 9 C6H5 >100 mM >100 mM 208.6 370.1 10 4-CH3-C6H4 >100 mM >100 mM 83.0 309.3 11 4-Br-C6H4 >100 mM >100 mM 353.3 140.7 12 2-I-C6H4 >100 mM >100 mM 45.4 643.7 13 2-Br-C6H4 >100 mM >100 mM 66.8 96.2 14 2-F-C6H4 >100 mM >100 mM 74.6 40.3 15 2-CF3-C6H4 >100 mM >100 mM 19.7 8.7 16 thien-2-yl >100 mM >100 mM 177.5 73.2 17 furan-2-yl >100 mM >100 mM 210.1 134.4 AAZ – 250 12 25 5.7 a

Mean from three different assays, by a stopped flow technique (errors were in the range of ±5–10% of the reported values).

b Incubation time 6 h. O SO2 O2N O OH O2N OH O2N O O2N S O O O SO2 H2N 1 2 4 6 7 O SO2 H N O R 8-17 (i) SO2Cl 3 5 (iii) (iv) (ii) R= Me ; ; ; ; 8 (70%) 9 (72%) 10 (73%) 11 (59%) Me ; ; ; ; 12 (88%) 13 (82%) 14 (79%) 15 (87%) Br N N Ru Cl Cl PCy3 Ph 5 (v) Br I F F3C ; . 16 (81%) 17 (81%) S O

Scheme 1. Reagents and conditions: (i) MePPh3Br, tBuOK, THF, RT, 18 h, 65%; (ii) Net3, CH2Cl2, 0C to RT, 4 h, 57%; (iii)5, toluene, 70C, 4 h, 96%; (iv) Fe, AcOH,

EtOH, H2O, 75C, 1 h, 98%; (v) RCOCl, Net3, CH2Cl2, 0C to RT, 4 h.

(6)

excellent yields (see Experimental for details). The nature of moi-eties R was chosen in such a way to assure chemical diversity. Apart R¼ Me in compound 8, the remaining derivatives 9–17 incorporated aromatic or heterocyclic moieties, such as phenyl, 2-or 4-substituted phenyls, thienyl and furyl. We found out in previ-ous papers1–3 that aryl or hetaryl moieties on the sulfocoumarin, homosulfocoumarin or coumarin ring6 systems lead to com-pounds with an effective inhibition profile against CA isoforms of pharmacologic interest, such as the tumour-associated ones CA IX and XII.

3.2. Carbonic anhydrase inhibition

The obtained homosulfocoumarins 8–17 were investigated for their CA inhibitory properties by using a stopped-flow CO2 hydrase assay20and four human CA isoforms (hCA I, II, IX, and XII) known to be drug targets1(Table 1).

As seen from data ofTable 1, derivatives 8–17 did not signifi-cantly inhibit the cytosolic isoforms hCA I and II, similar to other homosulfocoumarins, sulfocouamrins or coumarins investigated earlier1–8. On the other hand, the transmembrane, tumour-associ-ated isoforms hCA IX and XII were inhibited by all these com-pounds in the nanomolar range. For hCA IX the KIs were in the range of 19.7–353.3 nM whereas for hCA XII in the range of 8.7–643.7 nM (Table 1). The nature of the R moiety on the carbox-amide functionality greatly influenced the inhibitory power. For hCA IX/XII the optimal substitution was that present in compound 15, 2-trifluoromethylphenyl, whereas the one leading to the least effective inhibitor was the one with 4-bromophenylcarboxamide moiety (compound 9) for hCA IX and 2-iodophenylcarboxamide (compound 12) for hCA XII. Overall, all these new homosulfocou-marins act as isoform IX/XII selective CAIs over hCA I and II, which is highly desirable for these new chemotypes with enzyme inhibi-tory properties.

4. Conclusions

A series of 3H-1,2-benzoxathiepine 2,2-dioxides incorporating 7-acylamino moieties were obtained by an original procedure start-ing from 5-nitrosalicylaldehyde which was treated with propenyl-sulfonyl chloride followed by cyclisation through a Wittig reaction of the bis-olefin intermediate. The new derivatives, belonging to the homosulfocoumarin chemotype, were assayed as inhibitors of the zinc metalloenzyme CA. Four pharmacologically relevant human (h) isoforms were investigated, the cytosolic hCA I and II, and the transmembrane, tumour-associated hCA IX and XII. No relevant inhibition of hCA I and II was observed, whereas some of the new derivatives were effective, low nanomolar hCA IX/XII inhibitors, making them of interest for investigations in situations in which the activity of these isoforms is overexpressed, such as hypoxic tumours, arthritis or cerebral ischaemia.

Disclosure statement

The author(s) do not declare any conflict of interest.

Funding

This work was partly supported by ERA.Net RUS plus joint pro-gramme project THIOREDIN (State Education Development Agency of Republic of Latvia) and the Russian Foundation for

Basic Research [project grant 18–515-76001] under ERA.Net RUS plus joint programme grant RUS_ST2017-309.

ORCID

Claudiu T. Supuran http://orcid.org/0000-0003-4262-0323

References

1. (a) Pustenko A, Stepanovs D, Zalubovskis R, et al. 3H-1,2-benzoxathiepine 2,2-dioxides: a new class of isoform-select-ive carbonic anhydrase inhibitors. J Enzyme Inhib Med Chem 2017;32:767–75.;(b) Pustenko A, Nocentini A, Balasova A, et al. Aryl derivatives of 3H-1,2-benzoxathiepine 2,2-diox-ide as carbonic anhydrase inhibitors. J Enzyme Inhib Med Chem 2020;35:245–54.

2. Tars K, Vullo D, Kazaks A, et al. Sulfocoumarins (1,2-benzoxa-thiine 2,2-dioxides): a class of potent and isoform-selective inhibitors of tumor-associated carbonic anhydrases. J Med Chem 2013;56:293–300.

3. Tanc M, Carta F, Bozdag M, et al. 7-Substituted-sulfocoumar-ins are isoform-selective, potent carbonic anhydrase II inhibi-tors. Bioorg Med Chem 2013;21:4502–10.

4. Nocentini A, Ceruso M, Carta F, Supuran CT. 7-Aryl-triazolyl-substituted sulfocoumarins are potent, selective inhibitors of the tumor-associated carbonic anhydrase IX and XII. J Enzyme Inhib Med Chem 2016;31:1226–33.

5. Grandane A, Tanc M, Mannelli LDC, et al. Substituted sulfo-coumarins are selective carbonic anhdydrase IX and XII inhibitors with significant cytotoxicity against colorectal can-cer cells. J Med Chem 2015;58:3975–83.

6. (a) Maresca A, Temperini C, Vu H, et al. Non-zinc mediated inhibition of carbonic anhydrases: coumarins are a new class of suicide inhibitors. J Am Chem Soc 2009;131:3057–62; (b) Maresca A, Temperini C, Pochet L, et al. Deciphering the mechanism of carbonic anhydrase inhibition with coumarins and thiocoumarins. J Med Chem 2010;53:335–44; (c) Temperini C, Innocenti A, Scozzafava A, et al. The coumarin-binding site in carbonic anhydrase accommodates structur-ally diverse inhibitors: the antiepileptic lacosamide as an example. J Med Chem 2010;53:850–4; (d) Touisni N, Maresca A, McDonald PC, et al. Glycosylcoumarin carbonic anhydrase IX and XII inhibitors strongly attenuate the growth of pri-mary breast tumors. J Med Chem 2011;54:8271–7.

7. Zengin Kurt B, Sonmez F, Durdagi S, et al. Synthesis, bio-logical activity and multiscale molecular modeling studies for coumaryl-carboxamide derivatives as selective carbonic anhydrase IX inhibitors. J Enzyme Inhib Med Chem 2017;32: 1042–52.

8. (a) Maresca A, Scozzafava A, Supuran CT. 7,8-disubstituted-but not 6,7-disubstituted coumarins selectively inhibit the transmembrane, tumor-associated carbonic anhydrase iso-forms IX and XII over the cytosolic ones I and II in the low nanomolar/subnanomolar range. Bioorg Med Chem Lett 2010;20:7255–8; (b) Maresca A, Supuran CT. Coumarins incorporating hydroxy- and chloro-moieties selectively inhibit the transmembrane, tumor-associated carbonic anhy-drase isoforms IX and XII over the cytosolic ones I and II. Bioorg Med Chem Lett 2010;20:4511–4.

9. (a) Xu Y, Feng L, Jeffrey PD, et al. Structure and metal exchange in the cadmium carbonic anhydrase of marine

(7)

diatoms. Nature 2008;452:56–61; (b) Del Prete S, Vullo D, Fisher GM, et al. Discovery of a new family of carbonic anhy-drases in the malaria pathogen Plasmodium falciparum-the g-carbonic anhydrases. Bioorg Med Chem Lett 2014;24: 4389–96; (c) Jensen EL, Clement R, Kosta A, et al. A new widespread subclass of carbonic anhydrase in marine phyto-plankton. ISME J 2019;13:2094–106.

10. (a) Capasso C, Supuran CT. Anti-infective carbonic anhydrase inhibitors: a patent and literature review. Expert Opin Ther Pat 2013;23:693–704; (b) Capasso C, Supuran CT. An over-view of the alpha-, beta- and gamma-carbonic anhydrases from Bacteria: can bacterial carbonic anhydrases shed new light on evolution of bacteria? J Enzyme Inhib Med Chem 2015;30:325–32; (c) Capasso C, Supuran CT. Bacterial, fungal and protozoan carbonic anhydrases as drug targets. Expert Opin Ther Targets 2015;19:1689–704; (d) Supuran CT, Capasso C. Biomedical applications of prokaryotic carbonic anhydrases. Expert Opin Ther Pat 2018;28:745–54; (e) Nishimori I, Minakuchi T, Morimoto K, et al. Carbonic anhy-drase inhibitors: DNA cloning and inhibition studies of the alpha-carbonic anhydrase from Helicobacter pylori, a new target for developing sulfonamide and sulfamate gastric drugs. J Med Chem 2006;49:2117–26.

11. (a) Supuran CT. Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nature Rev Drug Discov 2008;7:168–81; (b) Alterio V, Di Fiore A, D’Ambrosio K, et al. Multiple binding modes of inhibitors to carbonic anhydrases: how to design specific drugs targeting 15 differ-ent isoforms? Chem Rev 2012;112:4421–68; (c) Supuran CT. Structure and function of carbonic anhydrases. Biochem J 2016;473:2023–32; (d) Innocenti A, G€ulc¸in I, Scozzafava A, Supuran CT. Carbonic anhydrase inhibitors. Antioxidant poly-phenols effectively inhibit mammalian isoforms I-XV. Bioorg Med Chem Lett 2010;20:5050–3.

12. (a) Supuran CT. How many carbonic anhydrase inhibition mechanisms exist? J Enzyme Inhib Med Chem 2016;31: 345–60; (b) Nocentini A, Supuran CT. Advances in the struc-tural annotation of human carbonic anhydrases and impact on future drug discovery. Expert Opin Drug Discov 2019;14: 1175–97; (c) Supuran CT. Advances in structure-based drug discovery of carbonic anhydrase inhibitors. Expert Opin Drug Discov 2017;12:61–88; (d) De Simone G, Supuran CT. (In)organic anions as carbonic anhydrase inhibitors. J Inorg Biochem 2012;111:117–29.

13. (a) Supuran CT. Carbonic anhydrase inhibitors as emerging agents for the treatment and imaging of hypoxic tumors. Expert Opin Investig Drugs 2018;27:963–70; (b) Supuran CT. Carbonic anhydrase inhibitors and their potential in a range of therapeutic areas. Expert Opin Ther Pat 2018;28:709–12; (c) Supuran CT. Applications of carbonic anhydrases inhibi-tors in renal and central nervous system diseases. Expert Opin Ther Pat 2018;28:713–21; (d) Neri D, Supuran CT. Interfering with pH regulation in tumours as a therapeutic strategy. Nature Rev Drug Discov 2011;10:767–77; (e) Supuran CT, Alterio V, Di Fiore A, et al. Inhibition of carbonic anhydrase IX targets primary tumors, metastases, and cancer stem cells: three for the price of one. Med Res Rev 2018;38: 1799–836.

14. (a) Supuran CT. Carbonic anhydrases and metabolism. Metabolites 2018;8:25; (b) Supuran CT. Carbonic anhydrase inhibition and the management of hypoxic tumors. Metabolites 2017;7:E48; (c) Da’dara AA, Angeli A, Ferraroni M, et al. Crystal structure and chemical inhibition of

essential schistosome host-interactive virulence factor car-bonic anhydrase SmCA. Commun Biol 2019;2:333.

15. (a) Supuran CT, Ilies MA, Scozzafava A. Carbonic anhydrase inhibitors. Part 29. Interaction of isozymes I, II and IV with benzolamide-like derivatives. Eur J Med Chem 1998;33: 739–52; (b) K€ohler K, Hillebrecht A, Schulze Wischeler J, et al. Saccharin inhibits carbonic anhydrases: possible explanation for its unpleasant metallic aftertaste. Ang ew Chem Int Ed Engl 2007;46:7697–9; (c) Scozzafava A, Menabuoni L, Mincione F, et al. Carbonic anhydrase inhibi-tors: perfluoroalkyl/aryl-s ubstituted derivatives of aromatic/ heterocyclic sulfonamides as topical intraocular pressure-lowering agents with prolonged duration of action. J Med Chem 2000;43:4542–51; (d) Sent€urk M, G€ulc¸in I, Das¸tan A, et al. Carbonic anhydrase inhibitors. Inhibition of human erythrocyte isozymes I and II with a series of antioxidant phenols. Bioorg Med Chem 2009;17:3207–11.

16. (a) Supuran CT, Altamimi ASA, Carta F. Carbonic anhydrase inhibition and the management of glaucoma: a literature and patent review 2013-2019. Expert Opin Ther Pat 2019;29: 781–92; (b) Supuran CT. Carbon-versus sulphur-based zinc binding groups for carbonic anhydrase inhibitors? J Enzyme Inhib Med Chem 2018;33:485–95; (c) Bilginer S, Gul HI, Erdal FS, et al. Synthesis, cytotoxicities, and carbonic anhydrase inhibition potential of 6-(3-aryl-2-propenoyl)-2(3H)-benzoxa-zolones. J Enzyme Inhib Med Chem 2019;34:1722–9.

17. (a) Margheri F, Ceruso M, Carta F, et al. Overexpression of the transmembrane carbonic anhydrase isoforms IX and XII in the inflamed synovium. J Enzyme Inhib Med Chem 2016; 31:60–3; (b) Bua S, Di Cesare Mannelli L, Vullo D, et al. Design and synthesis of novel nonsteroidal anti-inflamma-tory drugs and carbonic anhydrase inhibitors hybrids (NSAIDs-CAIs) for the treatment of rheumatoid arthritis. J Med Chem 2017;60:1159–70; (c) Akgul O, Di Cesare Mannelli L, Vullo D, et al. Discovery of novel nonsteroidal anti-inflam-matory drugs and carbonic anhydrase inhibitors hybrids (NSAIDs-CAIs) for the management of rheumatoid arthritis. J Med Chem 2018;61:4961–77.

18. (a) Carta F, Di Cesare Mannelli L, Pinard M, et al. A class of sulfonamide carbonic anhydrase inhibitors with neuropathic pain modulating effects. Bioorg Med Chem 2015;23: 1828–40; (b) Supuran CT. Carbonic anhydrase inhibition and the management of neuropathic pain. Expert Rev Neurother 2016;16:961–8.

19. Di Cesare Mannelli L, Micheli L, Carta F, et al. Carbonic anhy-drase inhibition for the management of cerebral ischemia: in vivo evaluation of sulfonamide and coumarin inhibitors. Enzyme Inhib Med Chem 2016;31:894–9.

20. Khalifah RG. The carbon dioxide hydration activity of car-bonic anhydrase. I. Stop-flow kinetic studies on the native human isoenzymes B and C. J Biol Chem 1971;246:2561–73. 21. (a) Vermelho AB, da Silva Cardoso V, Ricci Junior E, et al.

Nanoemulsions of sulfonamide carbonic anhydrase inhibi-tors strongly inhibit the growth of Trypanosoma cruzi. J Enzyme Inhib Med Chem 2018;33:139–46; (b) Nocentini A, Carta F, Tanc M, et al. Deciphering the mechanism of human carbonic anhydrases inhibition with sulfocoumarins: compu-tational and experimental studies. Chemistry 2018;24: 7840–4; (c) Awadallah FM, Bua S, Mahmoud WR, et al. Inhibition studies on a panel of human carbonic anhydrases with N1-substituted secondary sulfonamides incorporating thiazolinone or imidazolone-indole tails. J Enzyme Inhib Med Chem 2018;33:629–38.

(8)

22. (a) Bua S, Bozdag M, Del Prete S, et al. Mono- and di-thio-carbamate inhibition studies of the d-carbonic anhydrase TweCAd from the marine diatom Thalassiosira weissflogii. J Enzyme Inhib Med Chem 2018;33:707–13; (b) Ferraroni M, Gaspari R, Scozzafava A, et al. Dioxygen, an unexpected car-bonic anhydrase ligand. J Enzyme Inhib Med Chem 2018;33: 999–1005; (c) El-Gazzar MG, Nafie NH, Nocentini A, et al. Carbonic anhydrase inhibition with a series of novel benze-nesulfonamide-triazole conjugates. J Enzyme Inhib Med Chem 2018;33:1565–74; (d) Akocak S, Lolak N, Bua S, Supuran CT. Discovery of novel 1,3-diaryltriazene sulfona-mides as carbonic anhydrase I, II, VII, and IX inhibitors. J Enzyme Inhib Med Chem 2018;33:1575–80.

23. (a) Nocentini A, Bonardi A, Gratteri P, et al. Steroids interfere with human carbonic anhydrase activity by using alternative binding mechanisms. J Enzyme Inhib Med Chem 2018;33: 1453–9; (b) Nocentini A, Trallori E, Singh S, et al. 4-Hydroxy-3-nitro-5-ureido-benzenesulfonamides selectively target the

tumor-associated carbonic anhydrase isoforms IX and XII showing hypoxia-enhanced antiproliferative profiles. J Med Chem 2018;61:10860–74; (c) Chohan ZH, Munawar A, Supuran CT. Transition metal ion complexes of Schiff bases. Synthesis, characterization and antibacterial properties. Met Based Drugs 2001;8:137–43; (d) Ozt€urk Sarikaya SB, Topal F, Sent€urk M, et al. In vitro inhibition of a-carbonic anhydrase isozymes by some phenolic compounds. Bioorg Med Chem Lett 2011;21:4259–62.

24. (a) Awadallah FM, Bua S, Mahmoud WR, et al. Inhibition studies on a panel of human carbonic anhydrases with N1-substituted secondary sulfonamides incorporating thiazoli-none or imidazolone-indole tails. J Enzyme Inhib Med Chem 2018;33:629–38; (b) Supuran CT, Clare BW. Carbonic anhy-drase inhibitors. Part 57. Quantum chemical QSAR of a group of 1,3,4-thiadiazole and 1,3,4-thiadiazoline disulfona-mides with carbonic anhydrase inhibitory properties. Eur J Med Chem 1999;34:41–50.

Riferimenti

Documenti correlati

L'affermazione giurisprudenziale secondo cui: «nel ca- so di coesistenza del processo esecutivo promosso sul- la base di un decreto ingiuntivo provvisoriamente esecutivo, del

L’esperienza maturata ha permesso di ottimizzare la tecnica di acquisizione e pre-processamento dei dati ALS in aree con morfologia complessa, il sensore sperimentato ha

Nessa linha, é importante salientar que o volume agora publicado tem a participação da da Faculdade de Odontologia de Valença e do Instituto Superior de Educação – ISE,

affatto la protezione dei dati personali. Sembra quasi che l'articolo 8 non sia stato affatto considerato nella stesura della proposta. Come per l'articolo 15 ECD, le tecnologie

nell’interlineo, sopra Luçía, la traduzione ha la glossa idest gratia cohoperans, debi- trice della chiosa latina di A 1 104. In questo caso possiamo osservare come il traduttore